Discovery, Synthesis, and Characterization of an Orally Bioavailable

Sep 17, 2013 - MLK3 regulates fMLP-stimulated neutrophil motility. Oksana Polesskaya , Christopher Wong , Luis Lebron , Jeffrey M. Chamberlain , Harri...
0 downloads 20 Views 630KB Size
Article pubs.acs.org/jmc

Discovery, Synthesis, and Characterization of an Orally Bioavailable, Brain Penetrant Inhibitor of Mixed Lineage Kinase 3 Val S. Goodfellow,*,† Colin J. Loweth,†,⊥ Satheesh B. Ravula,†,# Torsten Wiemann,† Thong Nguyen,† Yang Xu,∥,¶ Daniel E. Todd,§ David Sheppard,§ Scott Pollack,§ Oksana Polesskaya,‡ Daniel F. Marker,‡ Stephen Dewhurst,‡ and Harris A. Gelbard‡ †

Califia Bio Inc., 11575 Sorrento Valley Road, San Diego 92121, California, United States University of Rochester Medical Center, School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, New York 14642, United States § BioFocus, Chesterford Research Park, Saffron Walden, Essex CB10 1XL, U.K. ∥ Biofocus DPI, 9640 Towne Centre Drive, San Diego, California 92121, United States ‡

S Supporting Information *

ABSTRACT: Inhibition of mixed lineage kinase 3 (MLK3) is a potential strategy for treatment of Parkinson’s disease and HIV-1 associated neurocognitive disorders (HAND), requiring an inhibitor that can achieve significant brain concentration levels. We report here URMC-099 (1) an orally bioavailable (F = 41%), potent (IC50 = 14 nM) MLK3 inhibitor with excellent brain exposure in mouse PK models and minimal interference with key human CYP450 enzymes or hERG channels. The compound inhibits LPS-induced TNFα release in microglial cells, HIV-1 Tat-induced release of cytokines in human monocytes and up-regulation of phospho-JNK in Tatinjected brains of mice. Compound 1 likely functions in HAND preclinical models by inhibiting multiple kinase pathways, including MLK3 and LRRK2 (IC50 = 11 nM). We compare the kinase specificity and BBB penetration of 1 with CEP-1347 (2). Compound 1 is well tolerated, with excellent in vivo activity in HAND models, and is under investigation for further development.



INTRODUCTION We report here the discovery, synthesis, and characterization of URMC-099 (1), a new inhibitor of mixed lineage kinase type 3 (MLK3) with excellent blood−brain barrier penetration properties, which has shown neuroprotective and antineuroinflammatory properties in in vitro and in vivo models of HIV-1 associated neurocognitive disorders (HAND).1 Combination antiretroviral therapy (cART) has greatly increased both the life expectancy and quality of life for HIV-1 seropositive individuals and is one of the greatest success stories of modern drug development. However, as the population of AIDS patients has aged, it has become apparent that neurological impairments resulting from HIV infection are not controlled by cART and may indeed be exacerbated by some CNS penetrating antiretroviral agents used in HIV therapy.2 HAND encompasses a broad range of neurologic deficits that range from mild cognitive impairment to frank dementia and is the result of damage to normal synaptic architecture that is likely mediated by dysregulation of immune cells in the CNS. In the U.S., greater than 50% of AIDS patients experience some symptoms of HAND, with a significant percentage (15%) exhibiting neurologic morbidity severe enough to preclude normal activities of daily living with substantial economic impact for their healthcare.2 The hallmarks of HAND include: (1) a dysregulation of inflammatory cytokines and chemokines, (2) the recruitment of © XXXX American Chemical Society

monocytes to the CNS, (3) viral infection of microglia leading to interruption of their normal function, and (4) extensive synaptodendritic damage, which ultimately impacts polysynaptic pathways that are the substrate for HAND in affected regions of the brain. A host of inflammatory mediators have been implicated in cellular models of HAND, where TNF-α release and signaling likely play a major central role. A more limited subset of mediators has been identified as being upregulated in the cerebrospinal fluid (CSF) and post-mortem brain tissues of HAND patients. These mediators/effectors include TNFα, the chemokine monocyte chemoattractant protein (MCP-1), and from preclinical models, mixed-lineage kinase 3 (MLK3), an important control point in MAPK kinase regulated inflammation pathways.3 Mixed lineage kinases are mitogen activated protein kinase kinase kinases (MAPKKKs) with features of both serine− threonine and tyrosine kinases that regulate the c-Jun N-terminal kinase (JNK) mitogen activated protein kinase (MAPK) signaling cascade and also regulate p38 and extracellular signalregulated kinase (ERK).4−6 MLK3 (MAP3K11) is the most widely expressed MLK family member4−6 and is expressed in neurons7 (as well as other cell types).8,9 At the cellular level, MLK3 is activated by stress, including reactive oxygen species, Received: July 18, 2013

A

dx.doi.org/10.1021/jm401094t | J. Med. Chem. XXXX, XXX, XXX−XXX

Journal of Medicinal Chemistry

Article

significant CNS penetration, which might serve as a first-inclass treatment, adjunctive to cART, for HAND.

ceramide, and TNFα.10,11 At the molecular level, it is activated by Cdc42 and Rac, which interact with MLK3, and can cause it to dimerize via a leucine zipper interface, resulting in autophosphorylation at Thr277 and Ser281 within the protein activation loop and enzyme activation.12,13 HIV-1 Tat also leads to phosphorylation at these same residues in primary rat neurons14 and to activation of glycogen synthase kinase (GSK-3ß) in neurons.15,16 This is important because MLK3 can be activated as a result of direct phosphorylation by GSK-3β.17 Previously published MLK3 inhibitors, CEP-134718 (2), K252a6 (3), CEP-70119 (4), CEP-1100420 (5), and compound 621 (Figure 1), have been based largely on the protein kinase-



RESULTS AND DISCUSSION We have identified a 7-azaindole based MLK3 inhibitor 1, with potent in vitro activity against HIV-1 Tat mediated release of cytokines in human and mouse immune cells, including cultured mouse microglial BV-2 cells; it is also neuroprotective in mice exposed to HIV-1 Tat.1 In mouse brain imaging experiments, compound 1 has demonstrated its in vivo efficacy for preservation of normal synaptic architecture and reversal of neuroinflammation following CNS exposure to HIV-1 Tat.1 Because of a compelling profile of central anti-inflammatory effects and neuroprotection, compound 1 is being evaluated for adjunctive therapy with or without cART in humanized mice engrafted with human CD34 cells that can sustain peripheral and central infection with HIV-1.31 A proprietary focused library of kinase inhibitors from BioFocus Ltd. containing approximately 15000 unique compounds was screened against MLK3 in a radiometric filter plate assay format. A large number of active compounds with consistent structure activity themes were identified from structurally distinct chemical libraries. We focused our initial hit-to-lead development on the imidazopyrazinone series based around screening hit (7) (see Figure 3). This compound gave variable potency in enzyme inhibition assays (MLK3 IC50 ranged between 40−120 nM); we believed this variability was due to solubility issues. Cellular assays also suggested poor ability to penetrate cells (LPS-stimulated TNF-α release in BV-2 mouse microglial cells, IC50 = 23.6 μM). However, the simple structure, well-developed SAR from screening hits, ease of synthesis, and potential for scaffold hopping from this series of compounds more than compensated for its initial drawbacks. During the modification of this scaffold, the design of compounds for optimization focused on low molecular weight compounds (30

>30 >30 >30

3.7 4.2 13.7

>30 >30 >30

13.2 21.8 >30

>30 >30 >30

Figure 4. Inhibition of HIV Tat stimulated cytokine release in human monocytes with compound 1 at 100, 300, and 1000 nM concentrations (NT = no treatment).

findings were observed in human monocyte-derived macrophages (MDM, data not shown) with the most potent inhibition of our inhibitors against MCP-1 and TNF-α release from MDM. In Vivo Inhibition of HIV-1 Tat Induced JNK Signaling with Compound 1. HIV-1 Tat activates JNK in neurons and other cell types, via an MLK-dependent pathway.14,41−44 JNKs are major regulators of mammalian apoptotic cell death pathways and have also been proposed to have an important role in neurodegeneration.45 However, direct inhibition of JNK may not be an acceptable strategy for disease modification because the role of JNK in cell death is so ubiquitous in different cell types, tissues, and organs. Further, there is significant JNK phosphorylation and activity in neurons from healthy brain tissue, indicating that JNK plays multiple roles in the healthy CNS. A selective strategy of interfering with HIV-induced signaling upstream of JNK, such as MLK signaling, may provide an opportunity to develop a more selective treatment for HAND with less risk of side effects and undesired toxicity as well as a potential treatment for other neurodegenerative diseases.

Kinase inhibitors possessing amine-solubilizing groups such as the piperazine present potential risk for inhibition of hERG channels, with associated risks of QT prolongation. The automated Cerep whole cell patch-clamp technique37 (Qpatch 16, Sophion Biosciences) was used to record outward potassium currents from single cells transfected with the hERG channel. The IC50 for 1 was determined as 21 μM and was rated as low cardiac toxicity risk by the CEREP compound evaluation criteria. Inhibition of Tat-Induced Pro-inflammatory Cytokine Release by Primary Human Monocytes. We tested whether 1 could regulate the HIV-1 Tat-induced release of a set of pro-inflammatory cytokines from primary human monocytes. We focused on TNF-α, MCP-1, IL-6, and IL-8 because these are believed to be of importance of the pathogenesis of HAND.32−40 Compound 1 potently inhibited all of the mediators in a dose dependent fashion. Statistically significant inhibition, compared E

dx.doi.org/10.1021/jm401094t | J. Med. Chem. XXXX, XXX, XXX−XXX

Journal of Medicinal Chemistry

Article

based inhibitors cocrystallized in kinase active sites. Docking studies of compound 1 in the MLK1 ATP binding site were performed using the automated docking utilities contained within the molecular modeling suite MOE46 as well as manual dockings and minimizations, allowing complete flexibility of the protein and ligand (refer to Figure 6). MLK1 is an excellent

To verify in vivo inhibition of the HIV-1 Tat-induced JNK pathway activation by compound 1, C57BL/6 mice were injected intracerebrally with saline or HIV Tat1−72, 700 μm deep in somatosensory cortex by stereotactic injection. This paradigm reproducibly results in sustained in vivo neuroinflammation after Tat injection.1 Half of the mice received ip pretreatment/treatment with 10 mg/kg of compound 1, every 12 h, which continued until sacrifice, with a total of three doses before Tat injection and two doses post injection while the other half were untreated (n = 3 for each of the four groups). Mice received the Tat injection at 4−6 h after the third ip injection of compound 1. In separate single dose PK experiments, plasma levels measured at 4 h post injection of drug were 500 nM and brain levels were also at 500 nM. At 6 h post drug injection, plasma levels were at 275 nM and brain levels were at 233 nM. Mice were anesthetized and sacrificed 24 h after Tat injection. Levels of phosphorylated JNK (pJNK) were measured by Western blotting from whole brain lysates collected directly adjacent to the Tat injection site (the caudal medial quadrant of the left hemisphere). To control for variations in protein loading between experimental replicates, levels of pJNK were normalized to the expression of total α-tubulin (see Figure 5A). The untreated Tat exposed animals had a 75%

Figure 6. Potential binding site interactions analysis for MLK1 and compound 1 based on manual docking and minimization studies.

model for MLK3 binding, and 1 potently inhibits MLK1 (IC50 = 19 nM). In aggregate, these analyses suggested that the compound is likely a type 1 kinase inhibitor forming two hydrogen bonds to the hinge. The indole is likely positioned so that it may make potential key interactions with neighboring lysine, aspartic acid, asparagine, or phenylalanine residues. In these models, the piperazine moiety interacts with a critical arginine residue and is also partially solvent exposed. To the best of our knowledge, no X-ray crystal structure for MLK3 has yet been published. Cephalon has released an X-ray crystal structure (3DTC) of one of their inhibitors 6 cocrystallized in MLK1.22 All but one of the significant residues in the MLK1 ATP binding site are identical to MLK3 (Figure 6). Phe222, present in the hinge region of MLK1, is a tyrosine in MLK3, however, the side chain is outwardly facing and its impact is minimal on ligand binding. In our SAR studies, we observed that the pyrrolo NH and the pyridine nitrogen of the scaffold were required for potency. In addition, for the side chains, having an aromatic group such as an indole, aniline, or phenol was associated with high in vitro enzyme inhibition potency as they provided a hydrogen bond donor from the para position of an aromatic ring. In studying the sequences of related kinases, only MLK1, MLK3, and ZAP70 contained the Gly225-Gly226-Pro227 motif, positioned at the top of the hinge, however 10 kinases contained a proline residue at the position equivalent to Pro227. One such kinase was spleen tyrosine kinase, SYK, which had been crystallized with the Rigel compound, R406.47 In this structure, the 3,4,5-trimethoxyphenyl group extended into the

Figure 5. Intraperitoneal (ip) compound 1 treatment prevents HIV-1 Tat mediated increase in JNK phosphorylation in vivo in brain tissue ipsilateral to stereotactically injected Tat. (A) Immunoblots of the expression of pJNK and α-tubulin in brain tissue of mice stereotactically injected with saline (Sa) vehicle and no treatment or Sa+ systemic administration of 1 or stereotaxic injection of HIV-1 Tat and no treatment or stereotaxic injection of Tat with systemic administration of 1. (B) Densitometric analyses of the optical density of bands in (A), normalized to the expression of α tubulin in each experimental replicate. * = p < 0.05.

increase in the levels of phosphorylated JNK compared to the saline control animals (Figure 5B). Compound 1 treatment in the Tat exposed animals normalized phosphorylated JNK levels to those observed in saline controls (Figure 5B) (*, p < 0.05). We analyzed data depicted in Figure 5A,B by one-way ANOVA with Newman−Keuls post hoc test. Error bars indicate SEM, but the compound had no effect on pJNK levels in salinetreated animals. Molecular Modeling Studies with Compound 1. Analysis of the potential binding modes of compound 1 were based on known SAR as well as crystal structures of similar 7-azaindole F

dx.doi.org/10.1021/jm401094t | J. Med. Chem. XXXX, XXX, XXX−XXX

Journal of Medicinal Chemistry

Article

P38a, SRK, SYK, JNK1, and ZAP70; see Table 6). In this panel of important diverse kinases from TK and AGC families, only LCK, a fairly promiscuous kinase, yielded an IC50 potency below 500 nM. As interest in this compound progressed with uniformly superior activity in cellular and in vivo models of HAND, we began to explore reasons for the unique activity. Using a bioinformatics approach, we identified six kinases which have known high affinity for closely related azaindole structures for further screening of the inhibition profile of 1. Two of these kinases had crystal structures of bound 7-azaindole inhibitors available to allow us to explore modeling of kinase specificity based on the observed subsite interactions. Although compound 1 had initially appeared fairly selective on the original screening panel, on these six selected kinases compound 1 exhibited significant potency for several of the kinases in these biochemical assays for inhibition of phosphorylation of substrates (Table 7). We expanded our investigation

solvent exposed region where it was observed to buttress the proline and also interact with a lysine residue in the equivalent position to Arg203. (MLK3 numbering) This group was recognized within our SAR in early compounds such as 7 and 8, and it was proposed that a similar interaction could take place with this group extending into the solvent exposed region. The 7-azaindole scaffold, in recent years, has been demonstrated to be a privileged structure for forming interactions with the hinge region of ATP binding sites of several kinases, and these compounds are the subject of several patents and studies of binding fragments for several protein kinases including AKT1, IKKB, JAK3, SGK1, and ABL1 mutants.48 A binding mode analysis performed against kinase X-ray structures containing ligands with an azaindole scaffold revealed consistency with our binding mode hypothesis identified through docking studies with homology models and PDB 3DTC. Of particular interest to us were 7-azaindole ligands bound in PDB 3ETA (insulin-like growth factor-1 receptor (IGF-1R)) tyrosine kinase and 2Z60 (T315I mutant of ABL1), which exhibited a binding mode we had originally focused on from docking studies with homology models. In these structures, the pyrrolo hydrogen of the inhibitor donates a hydrogen bond to the carbonyl of the Hinge +1 residue, and the pyridine nitrogen accepts a hydrogen bond from the hinge +3 N−H moiety. 3-Position substituents would likely interact with Phe155 in MLK1 and Asn273. 5-Position substituents on the 7-azaindole scaffold may interact with Arg230 and may be largely solvent exposed, allowing the incorporation of solubilizing groups. This model is consistent with all structure−activity data. Protein Kinase Specificity. Previously, little protein kinase specificity data has been published for MLK3 inhibitors. Compound 3 is a fairly promiscuous inhibitor.49 Compound 4 was originally identified as a potent inhibitor of FLT3 or JAK isoforms but is also a fairly potent inhibitor of MLK kinases and a close analogue of 2.49 Compound 6 was reported to be a potent inhibitor of MLK1 and a weak inhibitor of TrkA and Fyn.21 Compound 1 (MLK3 IC50 = 14 nM) inhibits both MLK1 (IC50 = 19 nM) as well as MLK2 (IC50 = 42 nM) and the related MLK family member DLK (IC50 = 150 nM), as expected from the analysis of the active sites of these enzymes. We initially utilized a screen of nine protein kinases to profile potential compounds (AMPK, CDK1, ERK2, GSK3B, LCK,

Table 7. IC50 Valuesa for Kinase Panel Selected by Bioinformatics Analysis of Kinase Ligands compounds

kinase

IC50

kinase

IC50

1512 108 123 290 1125 1180 177 6290 39 >10,000 307 200 3280

LCK MEKK2 P38a ROCK1 ROCK2 SGK SGK1 SRC SYK TRKA TRKB ZAP70 ABL1

333 661 12050 1030 111 67 201 4330 731 85 217 5050 6.8

1

13

3 1180 4 591 257 200 560

285 720 9.5 330 288 1290 730

a Average of two determinations (nanomolar). IC50s for various kinases in specificity screens were conducted by Reaction Biology, Inc. (Malvern, Pennsylvania) as described in ref 51. MEF cell experiments described in Experimental Section.

of specificity using the binding kinome scan ScanMax assay from DiscoverRx (originally conducted at Ambit Biosciences), so comparison could be made with compounds described in the literature. In this assay, compound 1 was screened against a panel of 442 kinases and showed greater than 90% inhibition against 111 wild-type human protein kinases at 1 μM (data in Supporting Information). The ScanMax assay from DiscoverRx49,50 is widely used to compare kinase inhibitor specificity based on extrapolated kinase/inhibitor binding and to produce thermodynamic binding constants, Kd. The assay measures the ability of significantly modified kinases to bind to a solid support immobilized ligand and utilizes conditions not present in living cells, such as the absence of ATP. To confirm the results obtained with this assay, we therefore also conducted a scan of 342 human wild-type kinases for inhibition of substrate phosphorylation using a high throughput ATP-P33 radiolabeled assay from Reaction Biology Corp. at a dose of 1 μM in the presence of 10 μM ATP (data in Supporting Information).51 We followed up with individual radioligand binding data for ATP uptake to determine IC50 values for some of the more interesting kinases (see Tables 6 and 7). In the ScanMax binding format assay, at a concentration of 1 μM, compound 1 exhibited greater than 50% inhibition of binding for 265 kinases and showed greater than 99% inhibition of binding for 36 kinases. In the Reaction Biology format in a screen of 342 human wild-type kinases, compound 1 exhibited 50% or greater inhibition of 202 kinases, with 15 showing greater than 99% inhibition (see Supporting Information).

Table 6. IC50s (nM)a Determined for Selected Protein Kinases for Compound 1 AMPK AurA AurB AurC CDK1 CDK2 c-MET ERK2 FLT1 GSK3B IGF1R IR JNK1

kinase/assay ABL1 (T315I) CDK2 FLT3 IKKa IKKb IR FLT3(MEF Cells)

a

Average of two determinations (nanomolar). IC50s for various kinases in specificity screens were conducted by Reaction Biology, Inc. (Malvern, Pennsylvania) as described in ref 51. G

dx.doi.org/10.1021/jm401094t | J. Med. Chem. XXXX, XXX, XXX−XXX

Journal of Medicinal Chemistry

Article

Figure 7. Percent inhibition profiles in the DiscoverRx ScanMax kinome scan for compounds 1 and 2.

In the ScanMax binding format assay, at a concentration of 1 μM, 2 exhibited greater than 50% inhibition of binding of 185 kinases and showed greater than 99% inhibition of binding of 28 kinases including LRRK2, FLT3, CAMK1D, KIT, MLK1, and Aurora kinases A and C (see Figure 7 and Supporting Information). Overall, both compound 1 and 2 showed only modest kinase specificity with notable differences in selectivity. Compound 1 showed potent activity on the ABL1 family and important mutants, whereas 2 lacked potent activity in this family. In contrast, 2 showed high activity against certain CAMK family kinases, unlike compound 1. Activity against CAMK2A (84% inhibition for 2) and CAMK2D (87% inhibition for 2) is highly correlated with positive activity in micronucleus screening models for kinase induced chromosome damage, as are BRSK2 (94% inhibition) and AMPK-alpha2 (98% inhibition).53 Unlike 1, compound 2 also strongly interacted with a subset of AGC family kinases, inhibiting RSK1 and RSK4 and related kinases. Both compounds 1 and 2, as well as other MLK3 inhibitors not reported here, strongly interact with members of the STE family kinases (MAP4K4, TNIK, and MINK) involved in MAPK pathways. Our working hypothesis is that the remarkable effects observed in in vivo efficacy studies1 of 1 may be the result of compound 1 being a “selectively non-selective kinase inhibitor.” Indeed, recent strategies in kinase drug discovery now recognize that the most efficacious drugs modulate multiple kinase pathways, but a key issue remains in finding safe compounds with multiple activities.54 Two kinases, FLT3 and LRRK2, are both very potently inhibited by both compounds 1 and 2 and seem potentially interesting with regard to off-target and neuroprotective effects. FLT3 Inhibition. To date, every compound showing potent activity in our human monocyte assay for inhibition of cytokine induction by HIV-1 Tat have been potent inhibitors of FLT3 and MLK3. Moreover, a literature survey revealed that most

A large number of kinases showed very significant affinity for compound 1 in the ScanMax binding assay. These included the following important kinases that showed >90% inhibition at 1 μM: ABL1, CDK11, CDK4, CDKL2, CLK1, CLK2, CLK4, DYRK1B, FLT3, KIT, MELK, PDGFRB, SRPK2, ALK, ARK5, AXL, IKKalpha, IKKbeta, ROCK1, TYK2. Additionally, several kinases that are potential targets for antineurodegenerative kinase inhibitor programs showed substantial inhibition: DLK (88%, Kd = 70 nM, IC50 = 150 nM), LRRK2 (92%, IC50 = 11 nM), LRRK2 (G2019S mutant, 96%), MARK1 (92%), MARK2 (49%), IGF1R (48%), and SGK1 (70%). Compound 1 is essentially inactive against two kinases known to interact with MLK3 (GSK3β IC50 > 10 μM, as well as AKT1) and several other pathway-related or interesting kinases including AKT2, AKT3, ERK1, ERK2, p38 (all isoforms), BRAF, and EGFR. It is also noteworthy that 1 inhibited binding of JNK kinases potently in the ScanMax format, but that high JNK1, -2, and -3 activity was not confirmed in the biochemical assay measuring inhibition of substrate phosphorylation. For example, JNK1 showed 99% binding inhibition at 1 μM in the ScanMax assay, but the IC50 for phosphorylation inhibition was 3.2 μM. Comparison Kinome Scan of Compound 2. To define the specificity of 2 and to compare and contrast activity at potential control nodes for anti-inflammatory and neuroprotective pathways, we synthesized 252 as a standard and conducted kinase inhibition scans using the ScanMax assay to examine activity on 456 kinases, including most known human kinases and important mutants thereof. Compound 2 was characterized by 1H NMR, LC-MS, and high-resolution mass spectrometry, and the activity was confirmed in enzyme inhibition assays. As expected, 2 was a very potent inhibitor of the three major MLK enzymes with IC50s (average of two determinations) for MLK3 = 6 nM, MLK2 = 2 nM, MLK1 < 1 nM (refer to Experimental Section, method B). H

dx.doi.org/10.1021/jm401094t | J. Med. Chem. XXXX, XXX, XXX−XXX

Journal of Medicinal Chemistry

Article

Parkinson’s disease (which has been associated with the G2019S mutation in LRRK2) and Crohn’s disease.60 LRRK2 is potently inhibited by compound 1 (IC50 = 11 nM), and 2 also showed 100% inhibition of LRRK2 binding and 98% inhibition of the LRRK2 (G2019S) mutant binding in the ScanMax assay at a concentration of 1 μM. Interestingly, exposure of murine BV2 microglial cells to HIV-1 Tat results in phosphorylation of serine 935 on LRRK2 as well as release of pro-inflammatory cytokines and an increase in phagocytic activity. LRRK2 kinase inhibition attenuated Tatinduced cytokine release and phagocytosis, suggesting that LRRK2 may be a novel regulator of microglial inflammation in HAND.61 The role that kinases other than MLK3 play in the potent in vivo and in vitro activity of compound 1 in models of HAND is the subject of current investigation. Our hypothesis is that strong neuroprotective and antineuroinflammatory efficacy of compound 1 in our in vivo models for HAND are attributable to its ability to safely interfere with multiple kinase pathways that act cooperatively (or synergistically) to drive the pathogenesis of HAND. This has important implications for our understanding of HAND and suggests that cooperative kinaseregulated gene networks may play a critical and previously underappreciated role in this disease. The knowledge we have gained from the structure−activity relationships and requirements for metabolic stability and blood−brain barrier penetration have allowed us to design second-generation compounds not based on the 7-azaindole core that are much more selective for MLK3 and are potently neuroprotective in rodent cells, but the details of biological activity deviate surprisingly from those exhibited by 2 and compound 1; the details of these studies will be published in due course.

Table 8. Kinase Inhibitors with High Affinity for MLK3 and FLT357

a

kinase inhibitor

FLT3a

MLK3a

BIBF-1120 CEP-701 EXEL-2880 KW-2449 Pazopanib PD-173955 PKC-412 R406 Staurosporine TAE-684 VX-680

3.8 8.5 0.9 15 1100 150 11 0.71 2.9 15 6.5

700 18 340 230 740 550 17 11 20 420 680

ScanMax Binding Kd, nM.

previously described MLK3 inhibitors also potently inhibit FLT3 (Table 8). FLT3 is a cytokine/growth factor receptor tyrosine kinase and is important for survival, proliferation, and differentiation of hematopoietic cells in bone marrow. Inhibitors of FLT3 are being developed for the treatment of acute myeloid leukemia (AML)55 and are also of interest for chronic neuroinflammatory and neurodegenerative conditions such as multiple sclerosis.56 It is therefore notable that all of the azaindole MLK3 inhibitors disclosed here exhibited significant FLT3 activity (Table 9). Table 9. IC50 (Nanomolar) or Percent Inhibition at 1 μmol for FLT3 and LRRK2a

a

compd

FLT3

LRRK2

7 1 8 9 10 11 12 13

88% 4.0 98% 98% 99% 97% 98% 9.5

2970 11 35 46 5 ND ND ND



SUMMARY

Using potent and drug-like screening hits as a starting point, we employed a strategy to find CNS penetrant compounds based on low molecular weight, low polar surface area, limited number of hydrogen bond donors, and well-defined log D. By doing so, we identified a potent, orally bioavailable MLK3 inhibitor with excellent pharmacokinetic properties and improved CNS penetration over previously developed MLK3 inhibitors. This compound has excellent activity in preclinical models for HIV associated neurocognitive disease1 and is undergoing safety testing for potential development. Compound 1 potently inhibits several key kinases involved in multiple inflammatory and neurodegenerative pathways, including MLK3 and LRRK2, and is likely a ”selectively nonselective” kinase inhibitor which modulates cooperative kinase-regulated gene networks involved in the pathogenesis of HAND.

Average of two determinations.

Global suppression of FLT3 is not expected to be benign, and potent FLT3 inhibitors such as compound 4 have caused suppression of platelet production.58,59 However, in practice it is difficult to achieve cellular and in vivo inhibition of FLT3. We explored the cellular activity of our inhibitors in a cell-based FLT3 phosphorylation assay, using a murine embryonal fibroblast (MEF) cell line that expresses a high level of exogenous full-length human FLT3-wt. Stimulation of these cells with human FLT3-ligand results in FLT3 autophosphorylation. The IC50 for compound 1 against purified FLT3 was 4 nM, but in this cell-based assay, compound 1’s IC50 against FLT3 was 140fold less (560 nM). The activity of compound 13 was similar (730 nM). These findings suggest that our azaindole MLK3 inhibitors exhibit only modest levels of activity against FLT3 in cells and that FLT3 inhibition may not make a dominant contribution to the in vivo efficacy of compound 1. LRRK2 Inhibition. Leucine-rich repeat kinase 2 (LRRK2) represents another interesting kinase implicated in neurodegenerative and inflammatory diseases, including familial



CHEMISTRY Compounds were efficiently synthesized using sequential Suzuki couplings utilizing key intermediate 16. For routine synthesis of diverse analogues, best yields and purity were obtained by protecting the azaindole nitrogen as a tosyl group. The synthesis of compound 1 is illustrated in Scheme 1. 5-Bromo1H-pyrrolo[2,3-b]pyridine 14 was iodinated with NIS to afford 15. The N−H was protected using TsCl, and the resulting tosylate 16 was subjected to regioselective Suzuki coupling reaction using indole-5-boronic acid at room temperature to yield intermediate 17. The second Suzuki coupling with 4-formyl I

dx.doi.org/10.1021/jm401094t | J. Med. Chem. XXXX, XXX, XXX−XXX

Journal of Medicinal Chemistry

Article

Scheme 1. Synthesis of Compound 1a

a

Reagents and conditions: (a) NIS, acetone; (b) NaH, p-TSA, THF; (c) indole-5-boronic acid, PdCl2(PPh3)2, MeCN, aq 2 M Na2CO3; (d) 4-formylphenyl boronic acid, PdCl2(PPh3)2, MeCN, aq 2 M Na2CO3; (e) 1-methylpiperazine, Na(OAc)3BH, CH2Cl2; (f) aq 5N NaOH, CH2Cl2, acetone.

Scheme 2. Synthesis of Compound 13a

a Reagents and conditions: (a) 7-azaindole-5-boronic acid pinacol ester, PdCl2(PPh3)2, MeCN, aq 1 M Na2CO3; (b) NaH, p-toluene sulfonyl chloride, DMF; (c) 4-formylphenyl boronic acid, PdCl2(PPh3)2, MeCN, aq 1 M Na2CO3; (d) 1-methyl piperazine, Na(OAc)3BH, CH2Cl2; (e) NaOH, MeOH.

boronic acid with intermediate 17 gave the benzaldehyde 18. Reductive amination reaction with 1-methyl piperazine followed by hydrolysis of the tosyl group resulted in the desired product 1 in an overall efficient synthesis. Compounds 8, 9, 10, 11, and 12 were produced using the same synthetic approach. Synthesis of compound 13 proceeded by similar sequential Suzuki couplings but required the protection of the side chain azaindole nitrogen to achieve best yields and purity (Scheme 2). Suzuki coupling intermediate 16 with 7-aza-1H-indol-5ylboronic acid provided 20, which was then tosylated to yield 21. The second Suzuki coupling produced intermediate aldehyde 22, which was reductively aminated and deprotected to yield compound 13. Resynthesis of Screening Hit 7. Aminopyrazine 24 was brominated with NBS, and the resulting dibromo compound 25 underwent regioselective substitution with 5-aminoindole. Cyclization of 26 was accomplished by heating with carbonyl

diimidazole. Suzuki coupling of 27, using standard conditions with the 3,4,5-trimethoxy substituted boronic acid, provided compound 7 (Scheme 3).



EXPERIMENTAL SECTION

The structures of compounds synthesized in the examples below were confirmed using the following procedures. LC-MS/UV/ELS analysis was performed on instrumentation consisting of Shimadzu LC-10AD vp series HPLC pumps and dual wavelength UV detector, a Gilson 215 autosampler, a Sedex 75c evaporative light scattering (ELS) detector, and a PE/Sciex API 150EX mass spectrometer. Then 5.0 μL injections were performed for each sample on a Phenomenex Gemini 5 μm C18 column. Mobile phases consisted of 0.05% formic acid in both HPLC grade water (A) and HPLC grade acetonitrile (B). Then 5.0 μL injections were performed for each sample, using gradient elution from 5% B to 100% B in 4 min at a flow rate of 2.0 mL/min with a final hold at 100% B of 1.8 min. UV (220 and 254 nm) and ELS data was collected for 4.5 min. All final compounds exhibited >95% purity. Routine one-dimensional NMR spectroscopy was performed J

dx.doi.org/10.1021/jm401094t | J. Med. Chem. XXXX, XXX, XXX−XXX

Journal of Medicinal Chemistry

Article

Scheme 3. Synthesis of Screening Hit 7a

a

Reagents and conditions: (a) NBS, CH2Cl2, 0 °C; (b) EtOH, DIEA, 80 °C; (c) CDI, THF, 65 °C; (d) PdCl2(PPh3)2, CH3CN, H2O, 150 °C. bis(triphenylphosphine)-palladium(II) dichloride (6.0 mg, 0.008 mmol), and 1 M Na2CO3 (1 mL). The resulting mixture was degassed with Ar for 10 min, after which it was heated at 150 °C for 10 min in a Personal Chemistry Optimizer. The organic layer was separated, filtered, and concentrated in vacuo. The residue was purified by preparatory HPLC to yield 7 (6.5 mg, 19%). 1H NMR (DMSO-d6, 300 MHz): δ 12.18 (s, 1H), 11.28 (s, 1H), 8.57 (s, 1H), 7.83 (d, J = 1.8 Hz, 1H), 7.52 (d, J = 8.4 Hz, 1H), 7.42 (m, 1H), 7.37 (dd, J = 1.8, 8.4 Hz, 1H), 7.20 (s, 2H), 6.51 (m, 1H), 3.78 (s, 6H), 3.66 (s, 3H). HPLC retention time: 2.30 min. HR-MS (ESI-TOFMS) 418.1511 (M + H), calcd 418.1511. 3-(1H-Indol-5-yl)-5-(3,4,5-trimethoxyphenyl)-1H-pyrrolo[2,3-b]pyridine (8). 1H NMR (DMSO-d6, 300 MHz): δ 11.78 (s, 1H), 11.03 (s, 1 H), 8.51 (d, J = 2.1 Hz, 1H), 8.36 (d, J = 1.8 Hz, 1H), 7.86 (s, 1H), 7.72 (d, J = 2.4 Hz, 1H), 7.45 (s, 2H), 7.32 (m, 1H), 6.92 (s, 2H), 6.45 (m, 1 H), 3.85 (s, 6H), 3.70 (s, 3H). MS ESI (m/z): 400.4 (M + H)+, calcd 399. HRMS (EI) m/z calcd for C24H21N3O3Na (M + Na)+ 422.1475, found 422.1476. 5-(3-(1H-Indol-5-yl)-1H-pyrrolo[2,3-b]pyridin-5-yl)pyridin-2amine (9). 1H NMR (DMSO-d6, 300 MHz): δ 11.73 (d, J = 1.8 Hz, 1H), 11.05 (s, 1 H), 8.43 (d, J = 2.4 Hz, 1H), 8.29 (d, J = 1.8 Hz, 1H), 8.27 (d, J = 2.1 Hz, 1H), 7.88 (s, 1H), 7.76 (dd, J = 2.4, 8.4 Hz, 1H), 7.46 (s, 2H), 7.33 (m, 1H), 6.55 (dd, J = 0.6, 8.7 Hz, 1H), 6.46 (m, 1 H), 5.99 (s, 2 H). HPLC retention time: 1.10 min. MS ESI (m/z): 326.2 (M + H)+, calcd 325. HRMS (EI) m/z calcd for C20H16N5 (M + H)+ 326.1400, found 326.1401. 5-(5-(3,4,5-Trimethoxyphenyl)-1H-pyrrolo[2,3-b]pyridin-3yl)pyridin-2-amine (10). 1H NMR (DMSO-d6, 300 MHz): δ 11.82 (s, 1H), 8.53 (d, J = 1.8 Hz, 1H), 8.31 (d, J = 1.8, 1 H), 8.28 (d, J = 1.5 Hz), 7.76 (dd, J = 2.1, 8.4 Hz, 1 H), 7.70 (d, J = 2.4 Hz, 1H), 6.95 (s, 2H), 6.54 (d, J = 8.4 Hz, 1 H), 5.87 (s, 2H), 3.86 (s, 6H), 3.68 (s, 3H). MS ESI (m/z): 377.4 (M + H), calcd 376. HRMS (EI) m/z calcd for C21H21N4O3 (M + H)+ 377.1608, found 377.1607. 5-(3-(1H-Indol-5-yl)-1H-pyrrolo[2,3-b]pyridin-5-yl)pyrimidin2-amine (11). MS ESI (m/z): 327.2 (M + H), calcd 326. HRMS (EI) m/z calcd for C19H15N6 (M + H)+ 327.1353, found 327.1354. 5-(5-(3,4,5-Trimethoxyphenyl)-1H-pyrrolo[2,3-b]pyridin-3yl)pyrimidin-2-amine (12). MS ESI (m/z): 378.4 (M + H), calcd 377. HRMS (EI) m/z calcd for C20H20N5O3 (M + H)+ 378.1561, found 378.1563. 5-(5-(4-((4-Methylpiperazin-1-yl)methyl)phenyl)-1H-pyrrolo[2,3-b]pyridin-3-yl)-1H-pyrrolo[2,3-b]pyridine (13). To a solution of 23 (4.00 g, 5.46 mmol) in MeOH (20 mL) was added NaOH (723 mg, 16.4 mmol). The resulting mixture was heated at 50 °C for 60 min. After completion of the reaction, the product was extracted

on a 300 MHz Varian Mercury-Plus spectrometer. The samples were dissolved in deuterated solvents obtained from Cambridge Isotope Laboratories, Inc. and transferred to 5 mm ID NMR tubes. The spectra were acquired at 293 K. C, H, N, Pd combustion analysis provided by Robertson Mircrolit Laboratories, and high resolution mass spectra were obtained by the UCSD Department of Chemistry Agilent 6230 ESI-TOF mass spectrometer service. 3-(1H-Indol-5-yl)-5-(4-((4-methylpiperazin-1-yl)methyl)phenyl)-1H-pyrrolo[2,3-b]pyridine (1). In a round-bottom flask, 19 (12.4 g, 21.5 mmol) was dissolved in CH2Cl2 (105 mL) and MeOH (105 mL), then 5N NaOH (210 mL) added. The resulting mixture was heated at 50 °C for 60 min. After completion of the reaction, the product was extracted using EtOAc and purified on silica gel chromatography. After the purification, all combined batches were again dissolved in DCM (250 mL), MeOH (250 mL), and THF (250 mL) and added quadra pure TU thiourea resin (10 g) and stirred overnight on rotary evaporator. After filtration, the solvents were removed and redissolved in EtOH (450 mL) then heated to 55 °C. After 2 h, HPLC grade water was added and the resulting mixture kept in a freezer to afford white solid was collected by filtration to afford 3-(1Hindol-5-yl)-5-(4-((4-methylpiperazin-1-yl)methyl)phenyl)-1H-pyrrolo[2,3-b]pyridine 1 (6.4 g, 70%). 1H NMR (DMSO-d6, 300 MHz): δ 11.82 (bs, 1H), 11.07 (bs, 1H), 8.54 (d, J = 1.2 Hz, 1H), 8.41 (d, J = 1.2 Hz, 1H), 7.89 (s, 1H), 7.76 (d, J = 1.5 Hz, 1H), 7.69 (d, J = 5.1 Hz, 2H), 7.48 (m, 2H), 7.39 (d, J = 4.8 Hz, 2H), 7.35 (m, 1H), 6.49 (dd, J = 2.7, 1.2 Hz, 1H), 3.48 (s, 2H), 2.50 (m, 3H), 2.38 (m, 5H). ESI (m/z) 422 (M + H), calcd 421. HRMS (EI) m/z calcd for C27H28N5 (M + H)+ 422.2341, found 423.2329. CHN. (9S,10R,12R)-9,12-Epoxy-1H-diindolo[1,2,3-fg:3′,2′,1′-kl]pyrrolo[3,4-i][1,6]benzodiazocine-10-carboxylic Acid, 5,16-Bis[(ethylthio)methyl]-2,3,9,10,11,12-hexahydro-10-hydroxy-9methyl-1-oxo Methyl Ester (2). 2 was synthesized from K252a (LC Laboratories, Woburn, MA) according to the method of Kaneko.52 1H NMR (DMSO-d6, 300 MHz): δ 9.13 (s, 1H), 8.63 (s, 1H), 7.95 (s, 1H), 7.88 (d, J = 5.1 Hz, 1H), 7.84 (d, J = 5.1 Hz, 1H), 7.46−7.44 (m, 2H), 7.10 (dd, J = 3.0, 4.2 Hz, 1H), 6.33 (s, 1H), 5.02 (d, J = 10.2 Hz, 1H), 4.95 (d, J = 10.2 Hz, 1H), 3.98 (s, 2H), 3.94 (s, 2H), 3.92 (s, 3H), 3.38−3.35 (m, 1H), 2.50−2.46 (m, 4H), 2.13 (s, 3H), 1.99 (dd, J = 3.0, 8.4 Hz, 1H), 1.23 (t, J = 2.4 Hz, 6H). MS ESI (m/z): 616.3 (M + H)+, calcd 616.2. HRMS (EI) m/z calcd for C33H33N3O5Na (M + Na)+ 638.1754, found 638.1755. 1-(1H-Indol-5-yl)-6-(3,4,5-trimethoxyphenyl)-1H-imidazo[4,5-b]pyrazin-2(3H)-one (7). To a solution of 24 (27 mg, 0.08 mmol) in CH3CN (1 mL) in a Personal Chemistry microwave reaction vial was added 3,4,5-trimethoxyphenylboronic acid (17 mg, 0.08 mmol), K

dx.doi.org/10.1021/jm401094t | J. Med. Chem. XXXX, XXX, XXX−XXX

Journal of Medicinal Chemistry

Article

(8.9 g, 89 mmol) and sodium triacetoxyborohydride (14.2 g, 67.2 mmol). The reaction mixture was stirred for 1 h at room temperature, after which it was partitioned between CH2Cl2 and brine. The organic layer was separated, dried over MgSO4, and concentrated in vacuo. The crude product was purified on silica gel column to give 15 (17 g, 68% yield). 1H NMR (DMSO-d6, 300 MHz): δ 11.23 (bs, 1H), 8.70 (d, J = 1.2 Hz, 1H), 8.40 (d, J = 1.5 Hz, 1H), 8.09 (d, J = 0.9 Hz, 2H), 8.07 (s, 1H), 7. 98 (s, 1H), 7.71 (d, J = 5.1 Hz, 2H), 7.52 (s, 2H), 7.41 (m, 5H), 7.24 (d, J = 4.5 Hz, 1H), 7.16 (dd, J = 10.8, 4.5 Hz, 2H), 6.52 (dd, J = 2.7, 1.2 Hz, 1H), 3.53 (s, 2H), 2.57 (m, 3H), 2.34 (m, 5H) 2.29 (s, 3H). MS ESI (m/z): 576 (M + H)+, calcd 575. 5-(5-Bromo-1-tosyl-1H-pyrrolo[2,3-b]pyridin-3-yl)-1Hpyrrolo[2,3-b]pyridine (20). To a stirred suspension of 16 (60 g, 125 mmol) and 7-aza-1H-indol-5-ylboronic acid (22.2 g, 138 mmol) in CH3CN (625 mL) was added 1 M Na2CO3 (312 mL) followed by bis(triphenylphosphine)palladium(II) dichloride (4.4 g, 6.2 mmol). The resulting mixture was stirred at room temperature for 1 h. After complete consumption of starting materials, the mixture was extracted with EtOAc and evaporated to dryness in vacuo and it was dissolved in CH2Cl2 (50 mL), absorbed onto Celite, and dried. The residue was purified via silica gel chromatography using CH2Cl2 as the eluent to obtain 20 (38.6 g, 65% yield). 1H NMR (DMSO-d6, 300 MHz): δ 11.21 (bs, 1H), 8.52 (d, J = 1.2 Hz, 1H), 8.47 (d, J = 1.5 Hz, 1H), 8.13 (s, 1H), 8.05 (d, J = 5.1 Hz, 2H), 7.92 (s, 1H), 7.51 (d, J = 4.8 Hz, 1H), 7.46 (dd, J = 5.1, 1.2 Hz, 1H), 7.43 (d, J = 5.1 Hz, 1H), 7.40 (dd, J = 3.9, 1.8 Hz, 2H), 6.52 (dd, J = 2.7, 1.2 Hz,1 H), 2.33 (s, 3H). MS ESI (m/z): 466.2/468.2 (M + H)+, calcd 466. 5-(5-Bromo-1-tosyl-1H-pyrrolo[2,3-b]pyridin-3-yl)-1-tosyl1H-pyrrolo[2,3-b]pyridine (21). To a stirred solution of 20 (1.01 g, 2.17 mmol) in 20 mL of anhydrous DMF was added NaH [60% dispersion in mineral oil] (130 mg, 3.25 mmol). The reaction mixture was stirred for 20 min at room temperature, after which p-toluene sulfonyl chloride (538 mg, 2.82 mmol) was added. The resulting mixture was stirred at room temperature for 2 h. The mixture was evaporated to dryness and quenched with water. The crude product was mixed with EtOAc (100 mL) and extracted. The organic solution was dried and evaporated in vacuo. The residue was purified via silica gel chromatography eluting with 20% EtOAc in hexanes to obtain 21 as a white solid (1.21 g, 90%). 1H NMR (DMSO-d6, 300 MHz) δ 8.76 (d, J = 1.2 Hz, 1H), 8.56 (d, J = 1.2 Hz, 1H), 8.54 (d, J = 1.2 Hz, 1H), 8.45 (d, J = 1.5 Hz, 1H), 8.39 (s, 1H), 8.04 − 8.01 (m, 4H), 7.96 (d, J = 2.4 Hz, 1H), 7.43 (d, J = 4.5 Hz, 4H), 6.87 (d, J = 2.4 Hz, 1H), 2.34 (s, 6H). MS ESI (m/z): 622.4/624.1 (M + 1)+, calcd 621. 4-(1-Tosyl-3-(1-tosyl-1H-pyrrolo[2,3-b]pyridin-5-yl)-1Hpyrrolo[2,3-b]pyridin-5-yl)benzaldehyde (22),. To a solution of 21 (1.32 g, 2.12 mmol) in CH3CN (20 mL) in a round-bottom flask was added 4-formylphenylboronic acid (349 mg, 2.33 mmol), bis(triphenylphosphine)-palladium(II) dichloride (149 mg, 0.212 mmol), and 1 M Na2CO3 (20 mL). The resulting mixture was heated to reflux for 3 h and then cooled to room temperature. The precipitated product was filtered and dried. The organic layer was extracted with EtOAc and washed with brine and evaporated to dryness to afford more crude material. The filtered solid and crude material from evaporation were redissolved in CH2Cl2, absorbed on Celite, and purified on silica gel column chromatography to yield 22 as a white solid (822 mg, 60% yield). MS ESI (m/z): 647.2 (M + H)+, calcd 646. 5-(5-(4-((4-Methylpiperazin-1-yl)methyl)phenyl)-1-tosyl-1Hpyrrolo[2,3-b]pyridin-3-yl)-1-tosyl-1H-pyrrolo[2,3-b]pyridine (23). To a solution of 22 (3.35 g, 5.18 mmol) in CH2Cl2 (50 mL) was added 1-methylpiperazine (1.15 mL, 10.4 mmol) and sodium triacetoxyborohydride (2.19 g, 10.4 mmol). The reaction mixture was stirred for 1 h at room temperature, after which it was partitioned between CH2Cl2 and brine. The organic layer was separated, dried over MgSO4, and concentrated in vacuo. The crude product was purified on silica gel column to afford 23 (2.65 g, 70% yield). 1H NMR (DMSO-d6, 300 MHz): δ 8.83 (d, J = 1.2 Hz, 1H), 8.71 (d, J = 1.2 Hz, 1H), 8.52 (d, J = 1.5 Hz, 1H), 8.44 (d, J = 1.2 Hz, 1H), 8.36 (s, 1H), 8.07 (d, J = 5.1 Hz, 2H), 8.02 (d, J = 5.1 Hz, 2H), 7.96 (d, J = 2.4 Hz, 1H), 7.71 (d, J = 4.8 Hz, 2H), 7.43 (d, J = 5.1 Hz, 4H), 7.38 (d, J = 4.8 Hz, 2H),

using 250 mL of IPA:CHCl3 (1:3) and water. The organic portions were dried and concentrated in vacuo. The crude product was purified on a silica gel column to afford 13 (1.57 g, 68%). 1H NMR (DMSO-d6, 300 MHz): δ 11.96 (s, 1H), 11.63 (s, 1H), 8.61 (d, J = 1.5 Hz, 1H), 8.56 (d, J = 1.2 Hz, 1H), 8.41 (d, J = 0.9 Hz, 1H), 8.31 (d, J = 1.2 Hz, 1H), 7.89 (d, J = 1.5 Hz, 1H), 7.73 (d, J = 4.8 Hz, 2H), 7.49−7.48 (m, 1H), 7.40 (d, J = 4.8 Hz, 2H), 6.51−6.49 (m, 1H), 3.49 (s, 2H), 2.38−2.36 (m, 4H), 2.21 (s, 3H). ESI (m/z) 423.4 (M + H)+, calcd 422. HRMS (EI) m/z calcd for C26H27N6 (M + H)+ 423.2292, found 423.2295. 5-Bromo-3-iodo-1H-pyrrolo[2,3-b]pyridine (15). In a 3 L round-bottom flask, 5-bromo-1H-pyrrolo[2,3-b]pyridine (63g, 319 mmol) was dissolved in 1500 mL of acetone. To the stirred mixture was added NIS (79.1g, 351 mmol), and the resulting mixture was stirred for 1.5 h; the precipitated solid was collected by filtration and washed with cold acetone (400 mL) to afford 15 (89.8 g, 88% yield) as white solid. 1H NMR (DMSO-d6, 300 MHz) δ 12.35 (bs, 1H), 8.31 (d, J = 1.5 Hz, 1H), 7.86 (dd, J = 1.2 Hz, 0.3 Hz, 1 H), 7.80 (d, J = 1.5 Hz, 1H). MS ESI (m/z): 322/324 (M + H)+, calcd 323. 5-Bromo-3-iodo-1-tosyl-1H-pyrrolo[2,3-b]pyridine (16). To a stirred solution of 15 (45.0g, 139 mmol) in 700 mL of anhydrous THF cooled to 0 °C with an ice bath was added NaH [60% dispersion in mineral oil] (8.3 g, 208 mmol). The reaction mixture was stirred for 20 min at 0 °C, after which p-toluenesulfonyl chloride (29.1 g, 153 mmol) was added. The resulting mixture was stirred at 0 °C for 1.5 h, after confirmation of completion by LCMS and TLC, the reaction mixture was warmed to room temperature, evaporated to dryness, and quenched with water. The crude product was mixed with EtOAc (1000 mL) refluxed for 1 h, and then hexane (500 mL) was added to precipitate the product 16 (60 g, 90% yield) as a light-yellow powder. 1 H NMR (DMSO-d6, 300 MHz) δ 8.50 (d, J = 1.5 Hz, 1H), 8.21 (s, 1H), 8.00 (d, J = 4.8 Hz, 0.3 Hz, 2H), 7.98 (d, J = 1.5 Hz, 1H), 7.44 (dd, J = 4.8 Hz, 0.3 Hz, 2H), 2.34 (s, 3H). MS ESI (m/z): 477.0/479.0 (M + 1)+, calcd 477. 5-Bromo-3-(1H-indol-5-yl)-1-tosyl-1H-pyrrolo[2,3-b]pyridine (17). To a stirred suspension of 16 (60 g, 125 mmol) and 1H-indol-5ylboronic acid (22.2 g, 138 mmol) in CH3CN (625 mL) was added 1 M Na2CO3 (312 mL) followed by bis(triphenylphosphine)palladium(II) dichloride (4.4 g, 6.2 mmol). The resulting mixture was stirred at room temperature for 1 h. After complete consumption of starting materials, the mixture was extracted with EtOAc and evaporated to dryness in vacuo, it was dissolved in CH2Cl2 (50 mL), absorbed onto Celite, and dried. The residue was purified via silica gel chromatography using CH2Cl2 as the eluent to obtain 17 (38.6 g, 65% yield). 1H NMR (DMSO-d6, 300 MHz): δ 11.21 (bs, 1H), 8.52 (d, J = 1.2 Hz, 1H), 8.47 (d, J = 1.5 Hz, 1H), 8.13 (s, 1H), 8.05 (d, J = 5.1 Hz, 2H), 7.92 (s, 1H), 7.51 (d, J = 4.8 Hz, 1H), 7.46 (dd, J = 5.1, 1.2 Hz, 1H), 7.43 (d, J = 5.1 Hz, 1H), 7.40 (dd, J = 3.9, 1.8 Hz, 2H), 6.52 (dd, J = 2.7, 1.2 Hz,1 H), 2.33 (s, 3H). MS ESI (m/z): 466.2/468.2 (M + H)+, calcd 466. 4-(3-(1H-Indol-5-yl)-1-tosyl-1H-pyrrolo[2,3-b]pyridin-5-yl)benzaldehyde (18). To a solution of 17 (29.5 g, 63.3 mmol) in CH3CN (315 mL) in a round-bottom flask was added 4-formylphenylboronic acid (11.4 g, 76 mmol), bis(triphenylphosphine)-palladium(II) dichloride (4.4 g, 6.3 mmol), and 1 M Na2CO3 (160 mL). The resulting mixture was heated to reflux for 2.5 h. The reaction was cooled to room temperature; the precipitated product was filtered and dried. The organic layer was extracted with EtOAc and washed with brine and evaporated to dryness to afford more crude material. The filtered solid and crude and material from evaporation were redissolved in CH2Cl2, absorbed on Celite, and purified via silica gel chromatography using CH2Cl2 as the eluent to afford 18 (38.6 g, 65% yield). 1H NMR (DMSO-d6, 300 MHz): δ 11.21 (bs, 1H), 10.07 (s, 1H), 8.81 (d, J = 1.2 Hz, 1H), 8.53 (d, J = 1.2 Hz, 1H), 8.13 (s, 1H), 8.08 (d, J = 5.1 Hz, 2H), 8.02 (m, 5H), 7.53 (dd, J = 5.1 Hz, 2H), 7.45 (d, J = 5.1 Hz, 2H), 7.46 (dd, J = 3.0, 1.5 Hz, 1H), 6.52 (dd, J = 2.7, 1.2 Hz,1 H), 2.34 (s, 3H). MS ESI (m/z): 492 (M + H)+, calcd 491. 3-(1-Tosyl-1H-indol-5-yl)-5-(4-((4-methylpiperazin-1-yl)methyl)phenyl)-1H-pyrrolo[2,3-b]pyridine (19). To a solution of 18 (22 g, 44.8 mmol) in CH2Cl2 (448 mL) was added 1-methylpiperazine L

dx.doi.org/10.1021/jm401094t | J. Med. Chem. XXXX, XXX, XXX−XXX

Journal of Medicinal Chemistry

Article

6.87 (d, J = 2.4 Hz, 1H), 3.48 (s, 2H), 2.40 − 2.25 (m, 8H), 2.14 (s, 3H). MS ESI (m/z): 731.4 (M + H)+, calcd 730. 3,5-Dibromopyrazin-2-amine (25). To a stirred solution of aminopyrazine 24 (8.21 g, 86.4 mmol) in anhydrous methylene chloride (215 mL) cooled to 0 °C was added N-bromosuccinimide (32.3 g, 181 mmol) in portions over a 6 h period, during which time the temperature of the reaction was kept below 0 °C. The resulting mixture was stored at 4 °C overnight, after which it was stirred vigorously and quenched with H2O (100 mL). The organic layer was separated, after which it was washed with saturated aqueous NaHCO3, washed with brine, dried over MgSO4, filtered, and evaporated in vacuo to yield a residue that was triturated with 20% EtOAc in hexanes to yield 25 (10.3 g, 47%) as a yellow−brown powder. 1H NMR (CDCl3, 300 MHz) δ 8.02 (s, 1H), 5.05 (bs, 2H). HPLC retention time: 1.99 min. MS ESI (m/z): 252.0/254.0/256.2 (M + 1)+, calcd 251. 6-Bromo-N2-(1H-indol-5-yl)pyrazine-2,3-diamine (26). To a stirred suspension of 25 (3.48 g, 13.7 mmol) and 1H-indol-5-amine (2.00 g, 15.0 mmol) in EtOH (3.5 mL) was added diisopropylethylamine (2.60 mL, 15.0 mmol). The resulting mixture was stirred for 48 h at 80 °C, after which it was partitioned between EtOAc and H2O. The organic layer was separated, after which it was washed with brine, dried over Na2SO4, filtered, and evaporated in vacuo to yield a residue that was purified via silica gel chromatography eluting with 1:1 EtOAc:hexanes to yield 26 (1.75 g, 42%) as a red−brown solid. 1H NMR (DMSO-d6, 300 MHz): δ 10.98 (s, 1H), 8.22 (s, 1H), 7.83 (s, 1H), 7.31−7.28 (m, 3H), 7.19 (d, J = 8.7 Hz, 1H), 6.43 (s, 2H), 6.36 (s, 1H). HPLC retention time: 2.07 min. MS ESI (m/z): 304.2/306.2 (M + H)+, calcd 303. 6-Bromo-1-(1H-indol-5-yl)-1H-imidazo[4,5-b]pyrazin-2(3H)one (27). To a solution of 26 (0.450 g, 1.48 mmol) in THF (5 mL) was added carbonyldiimidazole (1.20 g, 7.40 mmol). The resulting mixture was heated at 65 °C for 48 h, after which it was concentrated in vacuo and partitioned between EtOAc and H2O. The organic layer was separated, dried over MgSO4, filtered, and concentrated in vacuo to yield a residue that was purified via silica gel chromatography eluting with EtOAc to yield 27 (0.20 g, 41%). HPLC retention time: 2.07 min. MS ESI (m/z): 330.2/332.2 (M + 1) +, calcd 329. LPS-Induced TNFα Release from Microglial BV2 Cells. A TNFα release assay in LPS-stimulated BV-2 cells was performed essentially as described.62 Briefly, mouse microglial BV-2 cells were treated with test compounds followed by LPS (100 ng/mL final concentration), and culture supernatants were harvested 8 h thereafter for TNFα ELISA. Screening PK (iv Dosing) in Mice. Three mice were used for each time point. Male C57/BL/6 mice were dosed iv (10 mg/mL) by tail vein injection of a solution of 2 mg/mL in solutions containing the indicated compound and vehicles (compounds 1, 9, 10, 11 and 13, 5% DMSO, 40% PEG-400, 55% saline; compounds 8 and 12, 5% DMSO, 40% PEG-400, 55% H2O containing 20% HP-βCD). Blood samples of approximately 0.30 μL were collected from each mouse (n = 3 mice per time point) by retro-orbital bleed while the animals were anesthetized with isoflurane. Blood samples were collected in tubes containing sodium heparin as the anticoagulant, predose and at 0.083, 0.25, 0.5, 1, 2, 4, 6, 8, and 24 h postdose. Samples were centrifuged within 1 h of collection and plasma was collected and stored at −20 °C until analysis. Total concentrations of the compound were determined by liquid chromatography−tandem mass spectrometry (LC-MS/MS), following plasma protein precipitation with acetonitrile and injection of the supernatant onto the column (XTerraMS C18, 5 μm, 4.6 mm × 50 mm). The LC system comprised an Agilent (Agilent Technologies Inc., USA) 1100 series liquid chromatography equipped with G1379A degasser, G1311A Quantpump, G1313A autosampler, and G1316A column oven. Mass spectrometric analysis was performed using an API4000 (triple-quadrupole) instrument from AB Inc. (Canada) with an ESI interface. The aqueous mobile phase was water with 0.1% formic acid, and the organic mobile phase was methanol with 0.1% formic acid. The lower and upper limits of quantitation of the assay were 2.5 and 5000 ng/mL based on known standards, respectively. Brains were collected from three different animals at each time point, rinsed with ice-cold saline,

weighed, and stored at −80 °C until analysis. For compound quantitation, mouse brains were homogenized in 5 volumes of water. The homogenates were extracted by protein precipitation with acetonitrile. LC-MS/MS analysis was conducted as described for the plasma. Brain homogenate concentrations were converted to brain concentrations for the calculations of brain to plasma ratios. Brain Penetration Comparison for Compounds 1, 2, and 13. Compounds were dissolved in 5% DMSO, 40% PEG-400, and 55% saline to yield a nominal concentration of 2 mg/mL (pH = 8) and were dosed at 10 mg/kg in C57BL/6 mice by tail vein injection. Samples were collected as described above at 30, 60, and 180 min postdose. Mice were anesthetized, sacrificed and blood collected by retro-orbital bleeds, and brains collected (3 time points over 24 h, 3 mice per time point). Concentration of compounds was determined in plasma and brain samples as described above. Detailed Oral PK Study for Compound 1 in C57/Bl/6 Mice. Intravenous dosing for %F determination: Male C57/BL/6 mice were dosed iv (2.5 mg/kg) by tail vein injection of a solution of 2 mg/mL of compound 1 dissolved in 5% DMSO, 40% PEG-400, and 55% saline. Blood samples of approximately 0.30 μL were collected from each mouse (n = 3 mice per time point) by retro-orbital bleed while the animals were anesthetized with isoflurane. Blood samples were collected in tubes containing sodium heparin as the anticoagulant, predose and at 0.083, 0.25, 0.5, 1, 2, 4, 7, and 24 h postdose. Samples were centrifuged within 1 h of collection and plasma was collected and stored at −80 °C until analysis. Oral Dosing for %F determination: Compound was dosed by oral gavage (10 mg/kg) as a suspension of 1 mg/mL of 1 in a solution of 0.5% hydroxypropyl methylcellulose and 0.4% Tween-80 in pH 7.4 PBS buffer. Blood samples were collected in tubes containing sodium heparin as the anticoagulant, predose and at 0.25, 0.5, 1, 2, 4 6, 8, and 24 h post dose. Total concentrations of the compound were determined by liquid chromatography−tandem mass spectrometry (LC-MS/MS) as described above. Stability in Human Liver Microsomes. Stability of the test compound was determined in human liver microsomes by Cerep Inc., using standard assay conditions of 1 μM concentration of the test compound. Compound concentration was determined by HPLC, and reported values are the average of duplicate values. Stability in Hepatocytes. Stability of the test compound was determined in cryopreserved human, cynomolgus monkey, and CD-1 mice hepatocytes, by Cerep Inc., using standard assay conditions of 1 μM concentration of the test compound. Compound concentration was determined by HPLC, and reported values are the average of tripicate values. CYP2C9, 2D6, and 3A4 Inhibition. Test compound was incubated with pooled human liver microsomes at 37 °C in 0.1 M Tris buffer, pH 7.4, and its effect on the metabolism of probe substrates for CYP enzymes determined (2D6, diclofenac; 3A4, dextromethorphan; 2C9, midazolam). The compound was tested at six concentrations ranging from 0.12 to 30 μM. Conditions of incubation in this assay have been optimized to maintain first-order reaction conditions and to minimize the potential for nonspecific binding of probe or study compound. Reactions were terminated with acetonitrile containing analytical internal standard (carbamazepine), and samples were centrifuged to remove microsomal protein and analyzed using optimized HPLC and MS conditions. The MS responses for the solvent control samples were taken as the 100% reference values against which the inhibition of metabolism was measured. IC50 values were calculated using a sigmoidal dose−response equation within GraphPad Prism. Inhibition of HIV Tat-Induced Cytokine Release in Primary Human Monocytes. This assay was performed essentially as described.14,63 Briefly, human monocytes were isolated from freshly collected whole blood using CD14 immunomagnetic beads (MiltenyiBiotec), plated in 24-well plates and incubated with the specified compounds at the indicated concentrations (100, 3000, 1000 nM); in control wells, no compound was added. Then 30 min later, HIV-1 Tat was added to a final concentration of 50 nM; the cells were incubated for 8 h (in control wells, nothing was added [NT]). Cell supernatants were then collected, centrifuged to remove debris, transferred to new M

dx.doi.org/10.1021/jm401094t | J. Med. Chem. XXXX, XXX, XXX−XXX

Journal of Medicinal Chemistry

Article

microcentrifuge tubes, and frozen at −20 °C. A Luminex bead array assay was then performed to quantitate the indicated chemokines and cytokines. Results were measured in triplicate or quadruplicate, and data are presented as mean values; error bars denote the standard deviation. Note that similar results were obtained with monocytes derived from multiple (n > 5) different donors, as well as in terminally differentiated monocyte-derived macrophages (data not shown). * = p < 0.05; one-way ANOVA with Bonferroni’s correction (when compared to Tat only control). Inhibition of JNK Phosphorylation in HIV-Tat Treated Mice. Wildtype C57BL/6 mice were either pretreated with three doses of 10 mg/kg Compound 1 spaced 12 h apart (n = 6) or were left untreated (n = 6). Treatment with compound 1 continued every 12 h throughout the duration of the experiment. Half of the mice (three pretreated with compound 1, three untreated) received a stereotactic injection of 3 μL of 3 μg/μL HIV-1 Tat1−72 in PBS into the somatosensory cortex at the coordinates 1.0 mm posterior to Bregma, 1.0 mm lateral left of Bregma, and 0.7 mm ventral to the pial surface. The remaining mice received an injection of 3 μL of sterile PBS at the same coordinates. A 35 gauge needle with a 10 μL Hamilton syringe controlled by a microsyringe pump (Micro4 World Precision Instruments) was used to perform the injections, which were delivered at a flow rate of 80 nL/min to minimize brain injury occurring as a result of injection pressure. The needle and syringes were coated with Sigmacote (Sigma SL-2) to prevent the Tat from sticking to the inside of the syringe. Then 24 hours after injection, the mice were sacrificed by pentobarbital overdose; animals were then briefly transcardially perfused with ice-cold saline, and the brains were removed, sectioned, and flash frozen on dry ice. The brain tissue of interest was homogenized in a glass homogenizer in a solution of 1X Tris-buffered saline pH 7.4 with 0.05% Tween and protease and phosphatase inhibitors (no. 161280; Thermo Fisher Scientific). The homogenate was spun down at 13000 rcf for 15 min to remove insoluble debris, and the lysis supernatant was collected. The protein concentrations were measured and normalized using a Bradford assay. Then 12 μg of protein sample was then mixed with loading dye, boiled for 5 min, and run on a 4% to 15% SDS-PAGE gel (BioRad 456−1086) at 100 V. The gel was transferred onto a nitrocellulose membrane at 100 V for 66 min on ice. The membranes were blocked in 5% milk in 1× Tris-buffered saline with 0.05% Tween (TBS-T) for 1 h at room temperature with shaking. The membranes were washed three times in TBS-T for 10 min a wash. The primary antibody against phosphorylated JNK (Cell Signaling 4668P) was applied overnight at 4 °C at a concentration of 1:2000 in TBS-T with 5% milk. The next day, the membranes were washed three times in TBS-T for 10 min a wash. The horseradish peroxidase (HRP)-conjugated secondary antibody (BioRad 170−6515) was applied at a concentration of 1:11000 in 5% milk TBS-T for 45 min at room temperature with shaking. The membranes were washed three times in TBS-T and enhanced chemiluminescence (ECL) substrate (Thermo 34076, Thermo 34080) was applied for 3 min. The membranes were developed on film (Thermo 34091). To control for variations in protein loading, the membranes were stripped (Millipore 2504) and then blocked in 5% milk TBS-T for 30 min. The mouse anti-α-tubulin (Sigma T5168) was applied in 5% milk TBS-T overnight with shaking. The process of washing, secondary application, and developing was repeated in order to obtain the α-tubulin loading control blot. The optical density measurements used for quantification were obtained using ImageJ. * = p < 0.05; one-way ANOVA with Bonferroni’s correction. Discover RX ScanMax Kinome Binding Scan. Compounds that bind the kinase active site directly (sterically) or indirectly (allosterically) prevent kinase binding to the immobilized ligand, and will reduce the amount of kinase captured on a solid support. Conversely, test molecules that do not bind the kinase have no effect on the amount of kinase captured on the solid support. Screening “hits” are identified by measuring the amount of kinase captured in test versus control samples by using a quantitative qPCR method that detects the associated DNA label to determine a Kd for ligand binding. Compounds 1 and 2 were tested at 1 μM concentration against a panel

of 442 known protein kinases. Data are presented as percent of control activity remaining. (0% indicates very tight binders, 100% indicates no binding). Complete data are presented in the Supporting Information. Reaction Biology Wild-Type Human Kinome Inhibition Scan. Compound 1 was tested against 342 wild-type human kinases for inhibition of protein phosphorylation using the Reaction Biology Inc. Hot Spot P33 radio binding assay.51 The compound was tested in single dose duplicate mode at a concentration of 1 μM . Control Compound was tested in 10 dose IC50 mode with 3-fold serial dilution starting at 20 μM. Reactions were carried out at 10 μM ATP. Complete data are available in the Supporting Information. Kinase Specificity Inhibition Data. Kinase inhibition IC50 values were determined from 10 point curves. IC50s for various kinases in specificity screens were conducted by Reaction Biology, Inc. (Malvern, Pennsylvania) as described.51 Radiometric Filter Plate MLK3 Assay (Method A). MLK3 (200 ng (130 nM), Dundee, DU8313) was incubated with 1 μM inactive MKK7b (Dundee, DU703) in the presence of 2 μM cold ATP (Km) and 0.5 μCi/assay 33P ATP and appropriate concentrations of compounds. After a 20 min incubation, the reactions were washed through filter plates and read on a scintillation counter. Biochemical Assay for the Inhibition of Kinase Activity for MLK3 (Method B). Myelin basic protein (20 μM final concentration) was dissolved in 20 mM Hepes (pH 7.5) containing 10 μM MgCl2, 1 μM EGTA, 0.02% Brij35, 0.02 mg/mL BSA, 0.1 μM Na3VO4, 2 mM DTT, and 1% DMSO. Activated MLK3 was added and mixed (20 nM final concentration), and inhibitors were added in DMSO. 33 P-ATP (specific activity 500 μCi/μL) was delivered into the reaction mixture to initiate the reaction (ATP concentration: 10 μM), and the mixture was incubated at room temperature for 20 min. Percent Activity was determined using a proprietary HOTSPOT microfluidic filter binding technology.51 FLT3 Cellular Assay. This was performed by Proqinase GMBH, Freiburg, Germany. Briefly, this assay uses a murine embryonal fibroblast (MEF) cell line, which expresses a high level of exogenously introduced full-length human, wild-type FLT3. Stimulation of these cells with human FLT3-ligand results in receptor tyrosine autophosphorylation. MEF-FLT3-wt cells were plated in DMEM supplemented with 10% FCS in multiwell cell culture plates. After serum-starvation overnight, cells were incubated with compounds in serum-free medium. After 90 min incubation at 37 °C, cells were stimulated with FLT3-L at 250 ng/mL for 5 min. Quantification of substrate phosphorylation was assessed in 96 well plates via sandwich ELISA using a substrate specific capture antibody and an antiphosphotyrosine detection antibody. Raw data were converted into percent substrate phosphorylation relative to controls (incubated with FLT3L alone), which were set to 100%. IC50 values were determined using GraphPad Prism 5.01 software using a nonlinear regression curve fit with variable hill slope. The equation is a four-parameter logistic equation. Plasma Protein Binding. Protein binding in human plasma was assessed by performing equilibrium dialysis with plasma containing test compounds (10 μM) against 0.1 M PBS (pH 7.4). Following incubation (6 h at 37 °C), the parent compound was measured in both plasma and buffer compartments by LC-MS and the percentage of compound bound to plasma proteins determined. Test compounds (10 μM) were added to plasma (n = 2) and dialyzed against 0.1 M phosphate buffered saline (pH 7.4) for 6 h at 37 °C. After incubation, the contents of each plasma and buffer compartment were removed and mixed with equal volumes of control buffer or plasma as appropriate to maintain matrix equivalence for analysis. Plasma proteins were then precipitated by the addition of acetonitrile containing carbamazepine as analytical internal standard, centrifuged, and the supernatant removed for analysis by mass spectrometry (LC-MS/MS). Molecular Modeling and Ligand Docking. The 3DTC.PDB structure of MLK1 was used as a surrogate for MLK3 to evaluate the binding mode for the series. The compounds contained a conserved scaffold that was hand docked into MLK1 and protein and ligand were N

dx.doi.org/10.1021/jm401094t | J. Med. Chem. XXXX, XXX, XXX−XXX

Journal of Medicinal Chemistry

Article

energy minimized using the molecular modeling suite MOE. It was hypothesized that the compounds acted as type 1 kinase inhibitor so the scaffold was positioned to ensure that the hydrogen bond donor and acceptor of the 7-azaindole core interacted with the corresponding contacts on the hinge. This provided two binding mode hypotheses for evaluation. The binding mode that fit the SAR data allowed the scaffold to donate and accept a hydrogen bond to and from Glu221 and Ala223, respectively. This placed the side-chain indole group to the rear of the ATP site with the N-methyl-piperazine group positioned toward the solubilizing pocket.



kinase 3 inhibitor URMC-099 is neuroprotective and anti-inflammatory in models of human immunodeficiency virus-associated neurocognitive disorders. J. Neurosci. 2013, 33, 9998−10010. (2) Heaton, R. K.; Franklin, D. R.; Ellis, R. J.; McCutchan, J. A.; Letendre, S. L.; Leblanc, S.; Corkran, S. H.; Duarte, N. A; Clifford, D. B.; Woods, S. P.; Collier, A. C.; Marra, C. M.; Morgello, S.; Mindt, M. R.; Taylor, M. J; Marcotte, T. D.; Atkinson, J. H.; Wolfson, T.; Gelman, B. B.; McArthur, J. C.; Simpson, D. M.; Abramson, I.; Gamst, A.; Fennema-Notestine, C.; Jernigan, T. L.; Wong, J.; Grant, I. HIVassociated neurocognitive disorders before and during the era of combination antiretroviral therapy: differences in rates, nature, and predictors. J NeuroVirol. 2011, 17, 3−16. (3) Gelbard, H. A.; Dewhurst, S.; Maggirwar, S. B.; Kiebala, M.; Polesskaya, O.; Gendelman, H. E. Rebuilding synaptic architecture in HIV-1 associated neurocognitive disease: a therapeutic strategy based on modulation of mixed lineage kinase. Neurotherapeutics 2010, 7, 392−398. (4) Gallo, K. A.; Johnson, G. L. Mixed-lineage kinase control of JNK and p38 pathways. Nature Rev. Mol. Cell. Biol. 2002, 9, 663−672. (5) Silva, R. M.; Kuan, C. Y.; Rakic, P.; Burke, R. E. Mixed lineage kinase-c-jun N-terminal kinase signaling pathway: a new therapeutic target in Parkinson’s disease. Mov. Disord. 2005, 20, 653−664. (6) Wang, L. H.; Besirli, C. G.; Johnson, E. M., Jr. Mixed-lineage kinases: a target for the prevention of neurodegeneration. Annu. Rev. Pharmacol. Toxicol. 2004, 44, 451−474. (7) Maroney, A. C.; Finn, J. P.; Connors, T. J.; Durkin, J. T.; Angeles, T.; Gessner, G.; Xu, Z.; Meyer, S. L.; Savage, M. J.; Greene, L. A.; Scott, R. W.; Vaught, J. L. Cep-1347 (KT7515), a semisynthetic inhibitor of the mixed lineage kinase family. J. Biol. Chem. 2001, 276, 25302−25308. (8) Handley, M. E.; Rasaiyaah, J.; Barnett, J.; Thakker, M.; Pollara, G.; Katz, D. R.; Chain, B. M. Expression and function of mixed lineage kinases in dendritic cells. Int. Immunol. 2007, 19, 923−933. (9) Handley, M. E.; Rasaiyaah, J.; Chain, B. M.; Katz, D. R. Mixed lineage kinases (MLKs): a role in dendritic cells, inflammation and immunity? Int. J. Exp. Pathol. 2007, 88, 111−126. (10) Jaeschke, A.; Davis, R. J. Metabolic stress signaling mediated by mixed-lineage kinases. Mol. Cell 2007, 27, 498−508. (11) Sathyanarayana, P.; Barthwal, M. K.; Kundu, C. N.; Lane, M. E.; Bergmann, A.; Tzivion, G.; Rana, A. Activation of the drosophila MLK by ceramide reveals TNF-alpha and ceramide as agonists of mammalian MLK3. Mol. Cell 2002, 10, 1527−1533. (12) Leung, I. W.; Lassam, N. Dimerization via tandem leucine zippers is essential for the activation of the mitogen-activated protein kinase kinase kinase, MLK-3. J. Biol. Chem. 1998, 273, 32408−32415. (13) Leung, I. W.; Lassam, N. The kinase activation loop is the key to mixed lineage kinase-3 activation via both autophosphorylation and hematopoietic progenitor kinase 1 phosphorylation. J. Biol. Chem. 2001, 276, 1961−1967. (14) Sui, Z.; Fan, S.; Sniderhan, L.; Reisinger, E.; Litzburg, A.; Schifitto, G.; Gelbard, H. A.; Dewhurst, S.; Maggirwar, S. B. Inhibition of mixed lineage kinase 3 prevents HIV-1 Tat-mediated neurotoxicity and monocyte activation. J. Immunol. 2006, 177, 702−711. (15) New, D. R.; Maggirwar, S. B.; Epstein, L. G.; Dewhurst, S.; Gelbard, H. A. HIV-1 Tat induces neuronal death via tumor necrosis factor-alpha and activation of non-N-methyl-D-aspartate receptors by a NFkappaB-independent mechanism. J. Biol. Chem. 1998, 273, 17852− 17858. (16) Dewhurst, S.; Maggirwar, S. B.; Schifitto, G.; Gendelman, H. E.; Gelbard, H. A. Glycogen synthase kinase 3 beta (GSK-3 beta) as a therapeutic target in neuroAIDS. J. Neuroimmune Pharmacol. 2007, 2, 93−96. (17) Mishra, R.; Barthwal, M. K.; Sondarva, G.; Rana, B.; Wong, L.; Chatterjee, M.; Woodgett, J. R.; Rana, A. Glycogen synthase kinase3beta induces neuronal cell death via direct phosphorylation of mixed lineage kinase 3. J. Biol. Chem. 2007, 282, 30393−30405. (18) Sweeney, Z. K.; Lewcock, J. W. ACS Chemical Neuroscience Spotlight on CEP-1347. ACS Chem. Neurosci. 2011, 2, 3−4.

ASSOCIATED CONTENT

S Supporting Information *

Kinase inhibition profiles for 442 kinases for compound 1 and 2 performed by DiscoverRx; inhibition profile using radio ligand binding assay for 342 wild-type human kinases performed by Reaction Biology Corp for compound 1. This material is available free of charge via the Internet at http://pubs.acs.org.



AUTHOR INFORMATION

Corresponding Author

*Phone: 858-720-1948. E-mail: vsgoodfellow@califiabio.com. Present Addresses ⊥

C.J.L.: Dart NeuroScience LLC, 7473 Lusk Boulevard, San Diego, California 92121, United States. # S.B.R.: Epigen Biosciences 10225 Barnes Canyon Road, San Diego, California 92121, United States. ¶ Y.X.: Ropes and Gray LLP, 1900 University Avenue, East Palo Alto, California 94303, United States. Author Contributions

The manuscript was written through contributions of all authors. All authors have given approval to the final version of the manuscript. Notes

The authors declare no competing financial interest.



ACKNOWLEDGMENTS We thank John Harris, John McCall, Angus MacLeod, and Sanjay B. Maggirwar for helpful guidance for this project and the National Institutes of Health for continuing funding of these efforts. This work was funded in part by grants from the National Institutes of Health, Division of Mental Health, NIH grant P01MH064570 (H.A.G., PI), and a Qualifying Therapeutic Discovery Project Grant to Califia Bio, Inc.



ABBREVIATIONS USED HAND, HIV-1-associated neurocognitive disorders; HIV-1, human immunodeficiency virus type 1; LPS, lipopolysaccharide; TNF-α, tumor necrosis factor α; MLK, mixed lineage kinase; CNS, central nervous system; BBB, blood−brain barrier; CSF, cerebrospinal fluid; hERG, human ether-à-gogo-related gene; JNK, c-Jun N-terminal kinase; FLT3, fmsrelated tyrosine kinase 3; LRRK2, leucine-rich repeat kinase 2; MOE, Molecular Operating Environment; NIS, N-iodosuccinimide; MeCN, CH3CN, acetonitrile; DIEA, di-isopropylethylamine; THF, tetrahydrofuran; CDI, 1,1′-carbonyldiimidazole; EtOAC, ethyl acetate; DMSO, dimethylsulfoxide; CH2CL2, DCM, dichloromethane; PK, pharmacokinetic



REFERENCES

(1) Marker, D. F.; Tremblay, M. E.; Puccini, J. M.; Barbieri, J.; Gantz Marker, M. A.; Loweth, C. J.; Muly, E. C.; Lu, S. M.; Goodfellow, V. S.; Dewhurst, S.; Gelbard, H. A. The new small-molecule mixed-lineage O

dx.doi.org/10.1021/jm401094t | J. Med. Chem. XXXX, XXX, XXX−XXX

Journal of Medicinal Chemistry

Article

(19) Davis, M. I.; Hunt, J. P.; Herrgard, S.; Ciceri, P.; Wodicka, L. M.; Pallares, G.; Hocker, M.; Treiber, D. K.; Zarrinkar, P. P. Comprehensive analysis of kinase inhibitor selectivity. Nature Biotechnol. 2011, 29, 1046−1051. (20) Falsig, J.; Pö rzgen, P.; Lotharius, J.; Leist, M. Specific modulation of astrocyte inflammation by inhibition of mixed lineage kinases with CEP-1347. J. Immunol. 2004, 173, 2762−2770. (21) Hudkins, R. L.; Diebold, J. L.; Tao, M.; Josef, K. A.; Park, C. H.; Angeles, T. S.; Aimone, L. D.; Husten, J.; Ator, M. A.; Meyer, S. L.; Holskin, B. P.; Durkin, J. T.; Fedorov, A. A.; Fedorov, E. V.; Almo, S. C.; Mathiasen, J. R.; Bozyczko-Coyne, D.; Saporito, M. S.; Scott, R. W.; Mallamo, J. P. Mixed-lineage kinase 1 and mixed-lineage kinase 3 subtype-selective dihydronaphthyl [3,4-a]pyrrolo[3,4-c]carbazole-5ones: optimization, mixed-lineage kinase 1 crystallography, and oral in vivo activity in 1-methyl-4-phenyltetraphydropyridine models. J. Med. Chem. 2008, 51, 5680−5689. (22) Lotharius, J.; Falsig, J.; van Beek, J.; Payne, S.; Dringen, R.; Brundin, P.; Leist, M. Progressive degeneration of human mesencephalic neuron-derived cells triggered by dopamine-dependent oxidative stress is dependent on the mixed-lineage kinase pathway. J. Neurosci. 2005, 25, 6329−6342. (23) Saporito, M. S.; Brown, E. M.; Miller, M. S.; Carswell, S. CEP1347/KT-7515, an inhibitor of c-jun N-terminal kinase activation, attenuates the 1-methyl-4-phenyl tetrahydropyridine-mediated loss of nigrostriatal dopaminergic neurons in vivo. J. Pharmacol. Exp. Ther. 1999, 288, 421−427. (24) Mathiasen, J. R.; McKenna, B. A.; Saporito, M. S.; Ghadge, G. D.; Roos, R. P.; Holskin, B. P.; Wu, Z. L.; Trusko, S. P.; Connors, T. C.; Maroney, A. C.; Thomas, B. A.; Thomas, J. C.; Bozyczko-Coyne, D. Inhibition of mixed lineage kinase 3 attenuates MPP+-induced neurotoxicity in SH-SY5Y cells. Brain Res. 2004, 1003, 86−97. (25) Bodner, A.; Toth, P. T.; Miller, R. J. Activation of c-Jun Nterminal kinase mediates gp120IIIB- and nucleoside analogue-induced sensory neuron toxicity. Exp. Neurol. 2004, 188, 246−253. (26) Bodner, A.; Maroney, A. C.; Finn, J. P.; Ghadge, G.; Roos, R.; Miller, R. J. Mixed lineage kinase 3 mediates gp120IIIB-induced neurotoxicity. J. Neurochem. 2002, 82, 1424−1434. (27) Eggert, D.; Dash, P. K.; Gorantla, S.; Dou, H.; Schifitto, G.; Maggirwar, S. B.; Dewhurst, S.; Poluektova, L.; Gelbard, H. A.; Gendelman, H. E. Neuroprotective activities of CEP-1347 in models of neuroAIDS. J. Immunol. 2010, 184, 746−756. (28) Parkinson Study Group PRECEPT Investigators. Mixed lineage kinase inhibitor CEP-1347 fails to delay disability in early Parkinson disease. Neurology 2007, 69, 1480-1490. (29) Ma, Q.; Gelbard, H. A.; Maggirwar, S. B.; Dewhurst, S.; Gendelman, H. E.; Peterson, D. R.; Difrancesco, R.; Hochreiter, J. S.; Morse, G. D.; Schifitto, G. Pharmacokinetic interaction of CEP-1347 and atazanavir in HIV-infected patients. J. Neurovirol. 2013, 3, 254−60. (30) StarDrop: Guiding Decisions in Drug Discovery; Optibrium Ltd.: Cambridge, UK, 2013; http://www.optibrium.com/stardrop/. (31) Dash, P. K.; Gorantla, S.; Gendelman, H. E.; Knibbe, J.; Casale, G. P.; Makarov, E.; Epstein, A. A.; Gelbard, H. A.; Boska, M. D.; Poluektova, L. Y. Loss of neuronal integrity during progressive HIV-1 infection of humanized mice. J. Neurosci. 2011, 31, 3148−3157. (32) Genis, P.; Jett, M.; Bernton, E. W.; Boyle, T.; Gelbard, H. A.; Dzenko, K.; Keane, R. W.; Resnick, L.; Mizrachi, Y.; Volsky, D. J.; Epstein, L. G.; Gendelman, H. E. Cytokins and arachidonic metabolites produced during human immunodeficiency virus (HIV)infected macrophage−astroglia interactions: implications for the neuropathogenesis of HIV disease. J. Exp. Med. 1992, 176, 1703− 1718. (33) Wesselingh, S. L.; Power, C.; Glass, J. D.; Tyor, W. R.; McArthur, J. C.; Farber, J. M.; Griffin, J. W.; Griffin, D. E. Intracerebral cytokine messenger RNA expression in acquired immunodeficiency syndrome dementia. Ann. Neurol. 1993, 33, 576−582. (34) Tyor, W. R.; Glass, J. D.; Griffin, J. W.; Becker, P. S.; McArthur, J. C.; Bezman, L.; Griffin, D. E. Cytokine expression in the brain during the acquired immunodeficiency syndrome. Ann. Neurol. 1992, 31, 349−360.

(35) Wesselingh, S. L.; Glass, J.; McArthur, J. C.; Griffin, J. W.; Griffin, D. E. Cytokine dysregulation in HIV-associated neurological disease. Adv. Neuroimmunol. 1994, 4, 199−206. (36) Chico, L. K.; Van Eldik, L. J.; Watterson, D. M. Targeting protein kinases in central nervous system disorders. Nature Rev. Drug Discovery 2009, 8, 892−909. (37) Cardiac Toxicity hERG; Cerep: Celle L’Evescault, France, 2012; http://www.cerep.fr/cerep/users/pages/downloads/Documents/ Marketing/Pharmacology%20&%20ADME/Application%20notes/ Cardiac%20toxicity%20hERG-2012.pdf. (38) Weiss, J. M.; Nath, A.; Major, E. O.; Berman, J. W. HIV-1 Tat induces monocyte chemoattractant protein-1-mediated monocyte transmigration across a model of the human blood−brain barrier and up-regulates CCR5 expression on human monocytes. J. Immunol. 1999, 163, 2953−2959. (39) Gonzalez, E.; Rovin, B. H.; Sen, L.; Cooke, G.; Dhanda, R.; Mummidi, S.; Kulkarni, H.; Bamshad, M. J.; Telles, V.; Anderson, S. A.; Walter, E. A.; Stephan, K. T.; Deucher, M.; Mangano, A.; SBologna, R.; Ahuja, S. S.; Dolan, M. J.; Ahuja, S. K. HIV-1 infection and AIDS dementia are influenced by a mutant MCP-1 allele linked to increased monocyte infiltration of tissues and MCP-1 levels. Proc. Natl. Acad. Sci. U. S. A. 2002, 99, 13795−13800. (40) Mankowski, J. L.; Queen, S. E.; Clements, J. E.; Zink, M. C. Cerebrospinal fluid markers that predict SIV CNS disease. J Neuroimmunol. 2004, 157, 66−70. (41) Li, C. J.; Ueda, Y.; Shi, B.; Borodyansky, L.; Huang, L.; Li, Y. Z.; Pardee, A. B. Tat protein induces self-perpetuating permissivity for productive HIV-1 infection. Proc. Natl. Acad. Sci. U. S. A. 1997, 94, 8116−8120. (42) Ganju, R. K.; Munshi, N.; Nair, B. C.; Liu, Z. Y.; Gill, P.; Groopman, J. E. Human immunodeficiency virus tat modulates the Flk-1/KDR receptor, mitogen-activated protein kinases, and components of focal adhesion in Kaposi’s sarcoma cells. J. Virol. 1998, 72, 6131−6137. (43) Kumar, A.; Manna, S. K.; Khawan, S.; Aggarwal, B. B. HIV-Tat protein activates c-Jun N-terminal kinase and activator protein-1. J. Immunol. 1998, 161, 776−781. (44) Singh, I. N.; El-Hage, N.; Campbell, M. E.; Lutz, S. E.; Knapp, P. E.; Nath, A.; Hauser, K. F. Differential involvement of p38 and JNK MAP kinases in HIV-1 Tat and gp120-induced apoptosis and neurite degeneration in striatal neurons. Neuroscience 2005, 135, 781−790. (45) Davies, C.; Tournier, C. Exploring the function of the JNK (cJun N-terminal kinase) signaling pathway in physiological and pathological processes to design novel therapeutic strategies. Biochem. Soc. Trans. 2012, 40, 85−89. (46) Santiago, V.; Giorgio, C.; Stefano, M. Medicinal Chemistry and the Molecular Operating Environment (MOE): application of QSAR and molecular docking to drug discovery. Curr. Top. Med. Chem. 2008, 8, 1555−1572. (47) Braselmann, S.; Taylor, V.; Zhao, H.; Wang, S.; Sylvain, C.; Baluom, M.; Qu, K.; Herlaar, E.; Lau, A.; Young, C.; Wong, B. R.; Lovel, S.; Sun, T.; Park, G.; Argade, A.; Jurcevic, S.; Pine, P.; Singh, R.; Grossbard, E. B.; Payan, D. G.; Masuda, E. S. R406, an orally available spleen tyrosine kinase inhibitor blocks fc receptor signaling and reduces immune complex-mediated inflammation. J. Pharmacol. Exp. Ther. 2006, 319, 998−1008. (48) Bamborough, P.; Brown, M. J.; Christopher, J. A.; Chung, C.; Mellow, G. W. Selectivity of kinase inhibitor fragments. J. Med. Chem. 2011, 54, 5131−5143. (49) Karaman, M. W.; Herrgard, S.; Treiber, D. K.; Gallant, P.; Atteridge, C. E.; Campbell, B. T.; Chan, K. W.; Ciceri, P.; Davis, M. I.; Edeen, P. T.; Faraoni, R.; Floyd, M.; Hunt, J. P.; Lockhard, D. J.; Milanov, Z. V.; Morrison, M. J.; Pallares, G.; Patel, H. K.; Pritchard, S.; Wodicka, L. M.; Zarrinkar, P. P. A quantitative analysis of kinase inhibitor selectivity. Nature Biotechnol. 2008, 26, 127−132. (50) Fabian, M. A.; Biggs, W. H., 3rd; Treiber, D. K.; Atteridge, C. E.; Azimioara, M. D.; Benedetti, M. G.; Carter, T. A.; Ciceri, P.; Edeen, P. T.; Floyd, M.; Ford, J. M.; Galvin, M.; Gerlach, J. L.; Grotzfeld, R. M.; Herrgard, S.; Insko, D. E.; Insko, M. A.; Lai, A. G.; Lelias, J. M.; Mehta, P

dx.doi.org/10.1021/jm401094t | J. Med. Chem. XXXX, XXX, XXX−XXX

Journal of Medicinal Chemistry

Article

S. A.; Milanov, Z. V.; Velasco, A. M.; Wodicka, L. M.; Patel, H. K.; Zarrinkar, P. P.; Lockhart, D. J. A small molecule-kinase interaction map for clinical kinase inhibitors. Nature Biotechnol. 2005, 23, 329− 336. (51) Ma, H.; Deacon, S.; Horiuchi, K. The challenge of selecting protein kinase assays for lead discovery optimization. Expert Opin. Drug Discovery 2008, 3, 607−621. (52) Kaneko, M.; Saito, Y.; Saito, H.; Matsumoto, T.; Matsuda, Y.; Vaught, J. L.; Dionne, C. A.; Angeles, T. S.; Glicksman, M. A.; Neff, N. T.; Rotella, D. P.; Kauer, J. C.; Mallamo, J. P.; Hudkins, R. L.; Murakata, C. Neurotrophic 3,9-bis[(alkylthio)methyl]-and-bis(alkoxymethyl)-K-252a derivatives. J. Med. Chem. 1997, 40, 1863− 1869. (53) Olaharski, A. J.; Gonzaludo, N.; Bitter, H.; Goldstein, D.; Kirchner, S.; Uppal, H.; Kolaja, K. Identification of a kinase profile that predicts chromosome damage induced by small molecule kinase inhibitors. PLoS Comput. Biol. 2009, 5, e1000446. (54) Morphy, R. Selectively nonselective kinase inhibition: striking the right balance. J. Med. Chem. 2010, 53, 1413−1437. (55) Grunwald, M. R.; Levis, M. J. FLT3 inhibitors for acute myeloid leukemia: a review of their efficacy and mechanisms of resistance. Int. J. Hematol. 2013, 97, 683−694. (56) DeBoy, C. A.; Rus, H.; Tegla, C.; Cudrici, C.; Jones, M. V.; Pardo, C. A.; Small, D.; Whartenby, K. A.; Calabresi, P. A. FLT-3 expression and function on microglia in multiple sclerosis. Exp. Mol. Pathol. 2010, 89, 109−116. (57) Davis, M. I.; Hunt, J. P.; Herrgard, S.; Ciceri, P.; Wodicka, L. M.; Pallares, G.; Hocker, M.; Treiber, D. K.; Zarrinkar, P. P. Comprehensive analysis of kinase inhibitor selectivity. Nature Biotechnol. 2011, 29, 1046−1051. (58) Santos, F. P.; Kantarjian, H. M.; Jain, N.; Manshouri, T.; Thomas, D. A.; Garcia-Manero, G.; Kennedy, D.; Extrov, Z.; Cortes, J.; Verstovsek, S. Phase 2 study of CEP-701, an orally available JAK2 inhibitor, in patients with primary or post-polycythemia vera/essential thrombocythemia myelofibrosis. Blood 2010, 115, 1131−1136. (59) Olaharski, A. J.; Bitter, H.; Gonzaludo, N.; Kondru, R.; Goldstein, D. M.; Zabka, T. S.; Lin, H.; Singer, T.; Kolaja, K. Modeling bone marrow toxicity using kinase structural motifs and the inhibition profiles of small molecular kinase inhibitors. Toxicol. Sci. 2010, 118, 226−275. (60) Tsika, E.; Moore, D. J. Mechanisms of LRRK2-mediated neurodegeneration. Curr. Neurol. Neurosci. Rep. 2012, 12, 251−260. (61) Marker, D. F.; Puccini, J. M.; Mockus, T. E.; Barbieri, J.; Lu, S. M.; Gelbard, H. A. LRRK2 kinase inhibition prevents pathological microglial phagocytosis in response to HIV-1 Tat protein. J. Neuroinflammation 2012, 9, 261. (62) Kiebala, M.; Maggirwar, S. B. Ibudilast, a pharmacologic phosphodiesterase inhibitor, prevents human immunodeficiency virus1 Tat-mediated activation of microglial cells. PLoS One 2011, 6, e18633. (63) Sui, Z.; Sniderhan, L. F.; Schifitto, G.; Phipps, R. P.; Gelbard, H. A.; Dewhurst, S.; Maggirwar, S. B. Functional synergy between CD40 ligand and HIV-1 Tat contributes to inflammation: implication in HIV type 1 dementia. J. Immunol. 2007, 178, 3226−3236.

Q

dx.doi.org/10.1021/jm401094t | J. Med. Chem. XXXX, XXX, XXX−XXX